Abstract
Immunotherapy in the form of allogeneic stem cell transplantation (SCT) plays an instrumental role in the treatment of acute myeloid leukemia (AML), with non-transplant modalities of immunotherapy including checkpoint blockade now being actively explored. Here, we provide an overview of the graft versus leukemia (GVL) effect in AML as a window into understanding the prospects of AML immunotherapy. We explore the roles of various cell types in orchestrating anti-leukemic immunity, as well as those contributing to the unique immune suppressive state of myeloid diseases. We discuss specific approaches to engage the immune system, while noting the challenges of the AML antigen landscape and the barriers to immune modulation. We review the potential for immunomodulatory agents in combination with cellular therapies, donor lymphocyte infusion, and following SCT. Finally, to address the challenge of minimal residual disease (MRD) following chemotherapy, we propose combination epigenetic and immunotherapy for the eradication of MRD.
Keywords: immunotherapy, AML, antibodies, vaccines, T cells, neoantigen, checkpoint blockade
Introduction
Patients with high-risk AML have a poor prognosis. High-risk AML can be defined by both biologic and clinical variables, including age, cytogenetics, mutational profile, exposure to prior genotoxic chemotherapy, and response to chemotherapy[1–8]. Once any AML patient has failed first-line approaches, there remains no standard of care for patients lacking targetable mutations[9]. Even with these failures, until recently, our standard approach to upfront therapy for AML induction and consolidation had remained static for decades.
For high-risk AML patients, allogeneic SCT (alloSCT) offers the best opportunity at cure[1]. AlloSCT is the most fundamental form of immunotherapy, in which the donor immune system recognizes and eradicates residual AML in the graft-versus-leukemia (GVL) effect[10]. However, there are significant limitations for alloSCT in high-risk AML. AlloSCT has the best outcomes in first complete remission (CR1), and many patients with high-risk AML do not achieve a first remission[1]. AlloSCT carries significant risks of treatment-related morbidity and mortality. Although reduced intensity conditioning regimens have expanded the number of patients eligible for allogeneic SCT, patients with advanced age are still more likely than younger patients to have co-morbidities precluding them from transplant. Even for those AML patients who are candidates, relapse after allogeneic SCT remains a significant challenge [1,11]. Recent data have also demonstrated that the presence of any detectable AML prior to transplant, or minimal residual disease (MRD), portends a poor prognosis and a high rate of relapse following alloSCT. These MRD positive patients may have an equally poor outcome as those who enter transplant with active AML[12], and represent an important area of unmet need for novel therapeutics. Thus, there is ample opportunity to improve outcomes in this difficult patient population.
Allogeneic Stem Cell Transplantation and Mechanisms of the Immune Responses to AML
The immunotherapeutic benefit of alloSCT was recognized with the observation that patients with higher rates of GVHD had lower rates of relapse[13]. As it was known that allogeneic T cells were major drivers of GVHD, it was hypothesized that these cells could also drive the GVL effect [14]. T cells recognize tumor cells through binding of the T cell receptor (TCR) to cognate major histocompatibility complex proteins (in humans, human leukocyte antigen, HLA) which present tumor antigen on the cell surface (Figure 1). This binding, along with positive costimulatory signals, activates the T cell to kill a tumor cell. This interaction can be exploited in several ways. First, mismatch at both minor and major histocompatibility antigens can drive a beneficial antitumor response in which donor T cells recognize a tumor cell as “foreign” in the recipient [13,15]. In HLA-matched sibling alloSCT, this is accomplished by disparate minor polymorphic non-self peptides, or minor histocompatibility antigens. By looking at large data sets, investigators have gradually improved donor-recipient HLA-typing to avoid GVHD yet preserve GVL [16]. To date, the majority of our understanding of the cellular response against AML comes from the decades of experience and improvements in allogeneic stem cell transplantation.
Figure 1. Overview of cellular mediators against AML blasts and potential pathways to engage them in vivo.

The various effector cells including NK cells, macrophages, effector and regulatory T cells are shown. Antibodies targeting tumor associated antigens (TAAs) on AML blasts can be in their native form, as antibody drug conjugates (ADCs), or generated as bispecifics to engage effector T cells. Dendritic cells, one of the most potent antigen presenting cells, can process antigens endogenously or provided through vaccination strategies in order to expand tumor-specific T cells.
Mobilizing the cellular response against AML
Multiple investigators have attempted to more specifically target T cells against commonly overexpressed tumor antigens in AML. Initial attempts at enhancing the antigen-specific T cell response to AML were based on vaccines eliciting tumor-specific T cells [18,19]. The basis for this approach was to provide defined tumor associated antigens to professional antigen presenting cells (APCs), such as dendritic cells, helping promote antitumor T cells (Figure 1). These included vaccines directed against minor histocompatibility antigens (HA-1, HA-2)[20], proteinase 3 (PR3) [21], Wilms tumor protein (WT-1)[19,22], or tumor cells overexpressing GM-CSF[23]. Given the largely immunosuppressive environment found in many malignancies including AML, not all vaccine responses have been sufficient enough to lead to in vivo expansion of antigen-specific T cells and tumor regression [24,25]. AML presents unique challenges for immunotherapy in that it is disseminated throughout the body at diagnosis and lacks the tumor draining lymph nodes key for orchestrating an effective immune response. The systemic nature of AML appears to induce a unique state of immunologic tolerance, an effect that is not seen when the same tumor is given subcutaneously[26]. Part of the immunosuppressive environment of AML can also be explained by defects seen in dendritic cells, which are important in stimulating the adaptive immune response. Kline and colleagues have demonstrated a role for CD8⍺ plasmacytoid DCs in mouse models of AML which are capable of cross-presenting leukemia associated antigens to T cells [27,28]. Patients with AML show both quantitative and functional defects in circulating blood dendritic cells which could hamper optimal cross-presentation [29]. More recently, our group has characterized a depletion of plasmacytoid dendritic cells with increasing levels of disease, indicating that a similar mechanism may be operative in humans[30].
As opposed to what was initially hypothesized, AML patients appear to have adequate numbers of T cells at diagnosis[31]. These T cells, however, are phenotypically different in that they express significantly higher levels of activation markers such as CD25 and CD69. Similar to what is found in pre-clinical models these T cells could appear activated in the setting of recognizing AML antigens but undergo abortive tolerance in the absence of DC stimulation[32]. In humans, this is supported by recent data demonstrating markers of T cell exhaustion (e.g. PD-1, TIM3, LAG3) and senescence at diagnosis, leading to divergent outcomes post-induction between responders and non-responders. Deep molecular profiling of responders has demonstrated upregulation of costimulatory markers and T cell signaling pathways indicating restoration of the T cell response[33]. Others have also shown increased levels of the inhibitory receptor PD-1 on T cells at the time of AML relapse, with a shift from naïve to effector memory populations, both supporting antigen-stimulated cells. These studies, however, saw no diminished function compared to healthy controls when these cells received ex vivo stimulation with mitogens [34]. In addition, more recent work found an increased frequency of Tregs in bone marrow aspirates from patients with AML, particularly in patients with greater than one relapse[31]. Given increased frequency of Tregs and increased expression of both exhaustion/inhibitory markers at diagnosis and relapse, there has been significant interest in the possible use of immune checkpoint blockade alone or in combination following induction chemotherapy.
Multiple immune checkpoint receptors including PD-1, OX40, and ICOS are expressed on T-cells in bone marrow aspirates from patients with newly diagnosed and relapsed AML[31]. Co-expression of specific immune checkpoint proteins (TIM-3 and LAG-3) with PD-1 is associated with T-cell exhaustion, and TIM-3 and LAG-3 are co-expressed more frequently with PD-1 on T-cells isolated from AML than T-cells from healthy donors[31]. PD-L1 is also expressed on some AML cells, although levels of PD-L1 expression have been variable across studies[35–39]. The largest case series shows that PD-L1 expression is low or undetectable on most cases of primary AML and expressed at low levels in only 16.3% (20/123) of cases at diagnosis[39]. However, smaller series suggest that PD-L1 expression may increase on AML blast cells during treatment and following relapse[35,36,40], and PD-L1 expression on AML cells is negatively correlated with outcome[36–38]. Pre-clinical mouse models also point towards the PD-1/PD-L1 pathway in immune evasion by AML[42–45], leading to several trials of checkpoint blockade in AML and MDS discussed in more detail below [46,47].
If tissue resident T cells are present but in a non-functional state, an alternative approach could be to isolate tumor specific T cells from patients, expand them to large numbers ex vivo and then re-infuse them into the patient. First developed at the NCI, adoptive T cell therapy has been used in many different clinical settings [48]. T cells directed against PR3, WT-1, mutated nucleophosmin, and TERT have been examined in AML[21,49,50]. Alternatively, T cells can be genetically modified with tumor-specific TCRs and expanded ex vivo. However, given that tumor-specific TCRs depend on precise TCR-HLA interactions, they are not broadly applicable as even the most common HLA-directed therapies against HLA-A2*01 are only found in ~20-30% of Caucasians. Therefore, alternative methods of targeting T cells to AML cells are needed. Given the success of chimeric antigen receptor (CAR)-expressing T cells in other hematologic malignancies, several groups have worked to identify appropriate target antigens in AML. Recently, Perna and colleagues took a non-biased proteomics approach and identified several highly expressed surface antigens in AML that may serve as promising targets for CAR-T cells in AML[51]. This would be an advance over current strategies targeting more broadly expressed antigens such as CD33 or CD123[52]. While these studies have had some success in patients with AML, much work needs to be done to optimize this promising therapy for this disease.
Natural killer (NK) cells, which are part of the innate immune system, also play an important role in the immune response against AML[53]. Following cytotoxic induction or maintenance therapy, NK cells are impaired and less cytotoxic towards AML blasts[54]. NK cells sense tumor cells differently than T cells in that they are not “restricted” to recipient HLA. This “missing-self” hypothesis was first demonstrated in mice where NK cells were able to lyse leukemia cells lacking HLA molecules[55]. In fact, lack of this recognition drives a strong anti-tumor response and this mismatch can be optimized in the SCT setting. NK cell activity in AML is driven primarily by the balance of activating and inhibitory signals delivered to the NK cell after it encounters the tumor. These signals are primarily driven through the family of killer immunoglobulin receptors, or KIR[56]. In AML, the importance of this interaction was first recognized clinically by Ruggeri and colleagues who found that in AML patients undergoing SCT, donors expressing KIR receptors not recognizing HLA ligands in the host had improved disease free survival and lower rates of GVHD[57]. Given the role of donor NK cells in alloSCT, Miller and colleagues pioneered the use of allogeneic NK cell infusions into patients with hematologic malignancies. They found that adoptive transfer of haploidentical NK cells with IL-2 infusion led to complete remissions in 5 of 19 patients with poor-risk AML. The patients who responded had an alloreactive NK cell repertoire in the graft vs host direction[58]. Based off these data, a number of adoptive cellular approaches have aimed to optimize NK cells for the treatment of AML[59,60]. While there are several pathways to manipulate in the cellular response, it remains important to consider the negative regulators that naturally limit these processes as another point of therapeutic intervention.
Negative regulators of the immune response to AML
In AML, both myeloid derived suppressor cells (MDSCs) and regulatory T cells play pivotal roles[64]. MDSCs are derived from hematopoietic progenitor cells otherwise capable of giving rise to monocytes, macrophages, neutrophils or granulocytes. MDSCs can be induced by the cytokines GM-CSF, IL-1, IL-6, and IL-10, all of which readily exist in the tumor microenvironment[65]. There are currently no good strategies for targeting MDSCs within the tumor microenvironment, partly owing to our lack of understanding of this ill-defined population of cells. MDSCs have been shown to create excess reactive oxygen species (ROS) within the tumor microenvironment inhibiting tumor reactive CD8 T cells[66].
T regulatory cells (Tregs) are important cells in maintaining tolerance to self, and thus play a major role in cancer development and immune suppression. As mentioned above, Tregs are increased in patients with AML, especially in the relapsed setting[31]. Tregs come in two forms, natural or induced. Natural Tregs develop in the thymus in response to high affinity interactions with medullary thymic epithelial cells expressing self-antigens and account for 5-10% of peripheral blood CD4 T cells. Peripheral Tregs can be induced from CD4 T cells in response to cytokines, such as TGF-β or IL-2 induced during inflammation and express high levels of the checkpoint CTLA-4. Tregs help control autoreactive T cells and thereby curtail autoimmune responses. This inhibitory mechanism can be usurped in cancer to help promote tolerance to certain tumors, including AML[31]. Studies in mice have shown that AML-associated Tregs can also block CTL responses by modulation of the PD-1/PD-L1 axis and that Treg depletion can lead to eradication of AML following PD1 blockade[67–69]. Soiffer and colleagues conducted a phase I trial looking at the use of CTLA-4 based checkpoint blockade in alloSCT recipients after relapse with Hodgkins lymphoma, multiple myeloma, or AML (including leukemia cutis and myeloid sarcoma)[70]. In twenty-one patients treated at 10 mg/kg ipilimumab, the overall response rate was 33%. 42% (5/12) of patients with AML achieved a complete response, with some of the responses durable at 8 month follow-up. Examination of immune correlates revealed that peripheral blood T cells peaked at 4 weeks after initiation of ipilimumab. Interestingly, peripheral Tregs decreased, with the ratio of Treg/Teff cells decreased in up to 41% of the 15 patients analyzed. This is consistent with the previously described mechanism of CTLA-4 blockade, in that it not only “takes off the brake” of T cells but also depletes CTLA-4 expressing Tregs in an Fc-dependent manner[71]. As one would expect, CTLA-4 blockade following alloSCT does have toxicity. Post-transplant ipilimumab was associated with immune-related adverse events (irAE) typical for ipilimumab in 6/28 patients (21%), including pneumonitis, diarrhea/colitis, and ITP. These irAEs were generally reversible with steroids, although one patient died of grade 3 colitis and grade 4 pneumonitis. Post-transplant ipilimumab also led to worsening of acute and chronic GVHD. Another study found similar changes in peripheral T cell counts (increased T cells, decreased frequency of Tregs) following CTLA-4 blockade as well as irAEs without evidence of GVHD, however, at lower doses of 3 mg/kg[73]. These studies demonstrate the need for a better understanding of immune reconstitution following alloSCT to more specifically modulate anti-leukemia efficacy.
Vaccine therapy revisited
While vaccination can potentially induce both T- and B-cell mediated immunity, prior attempts to vaccinate against single AML antigens did not lead to durable remissions. Thus, approaches providing a polyclonal response targeting multiple patient-derived antigens were hypothesized to be more effective against resistance and/or relapse. More recently, new approaches have re-invigorated this field with impressive responses. Avigan and Rosenblatt evaluated the use of an autologous tumor vaccine in 17 patients with high risk AML following remission induction and consolidation chemotherapy[74]. The vaccine consists of pre-treatment AML blasts from patients fused with patient derived DCs creating a hybridoma. Following vaccination, antigen-specific CD4 and CD8 T cell responses were induced against tumor antigens MUC1, WT1, and NY-ESO1. Compared to traditional responses in chemotherapy only treated patients (15-20% 2 year survival), twelve of the 17 (71%) patients remain in remission 5 years following initial treatment. There are now ongoing multi-center trials testing this approach. Other groups have attempted alternative methods to individualize recombinant vaccines on a per-patient basis. Using massively parallel sequencing to identify unique antigens in melanoma patients, investigators generated peptide vaccines with up to 20 personalized neoantigens admixed with toll-like receptor (TLR) agonists[75]. While this proof-of-concept study was a major advance in the field of vaccinology, it has yet to be extrapolated to other “cold” tumors where neo-antigen burden is substantially lower, including AML. The labor-intensive nature of such individualized cellular or recombinant products also presents significant challenges in scaling this approach for multiple patients.
The humoral response to AML
While the role of cellular immunity in controlling AML has been well described, much less is known about the antibody response. Some early anecdotal studies reported AML remissions in patients with hypergammaglobulinemia in the setting of transfusion or infections[76]. Others have worked to profile serum of AML patients for tumor associated antigens[77]. More recently, Hazenberg and colleagues took a novel approach to identify antibodies in patients with AML. Although the graft-vs-leukemia effect was thought to be primarily driven by T cells, they asked the simple question of whether or not protective antibodies can also be identified in patients following a successful allogeneic stem cell transplant. To do this this, they screened serum from transplant patients with high-risk disease at diagnosis who had survived past 5 years. By screening patient sera against AML lysates and following identification of positive clones, antibodies were isolated from these B cell clones[78]. Interestingly, they identified antibodies to a shared spliceosome protein U5 snRNP200 which is aberrantly expressed on the cell surface in AML patients. Furthermore, these antibodies showed activity against AML cells both in vitro and in vivo. They have gone on to characterize many other targets they have identified which could provide a new repertoire of antibody-based therapeutics for AML[79]. These data provide strong evidence that in combination with enhanced T cell activity, protective antibodies could provide durable control against high risk AML.
Although identification of these AML-specific antibodies provide important new targets for novel therapies, investigators have been using recombinant antibody technology to target AML since the early 2000s. These include both recombinant IgGs alone, as antibody drug conjugates (ADCs), or more recently bi-specific T cell engagers (BITEs), each of which have been extensively reviewed elsewhere[80].
Checkpoint blockade in the treatment of AML
The use of immunotherapy has changed the way we approach many cancers. Many immunotherapeutic drugs target inhibitory receptors and signaling pathways such as PD-1/PD-L1 and CTLA-4 that regulate immune cell activation in response to antigen stimulation, an approach described as immune checkpoint blockade[81–84]. Although the efficacy of immune therapy in hematologic malignancy has been established for decades in the form of allogeneic stem cell transplantation, the development of immune checkpoint inhibitors in hematologic disease has lagged behind solid tumors[46,47,85]. One notable exception involves the remarkable activity of single-agent PD-1 blockade in multiply relapsed/refractory Hodgkin lymphoma, where nivolumab yielded an overall response rate of 87% and CR rate of 17%[46,86]. In diseases such as leukemia, early-phase results from checkpoint inhibitor monotherapy have been less robust but this continues to be an evolving landscape[87]. One approach to improve response rates has been to combine multiple immune checkpoint inhibitors, though at the cost of increased toxicity from immune-related adverse events (irAEs)[82]. Another potential barrier to efficacy of checkpoint blockade is that de novo AML carries few mutations[88,89]. Mutational burden likely plays an important role in response to checkpoint blockade in promoting a specific neo-antigen landscape[90–92], thus ways to improve upon the recognition of AML blasts by a patient’s immune system are needed.
Epigenetic Therapy as Immmunotherapy
Epigenetic modifying drugs such as azacitidine and decitabine are commonly used in the treatment of patients with AML. They inhibit DNA methyltransferases and serve as hypomethylating agents (HMAs), altering patterns of gene expression. Although their mechanisms of action are not fully understood, HMAs provide some survival benefits to specific populations of AML and high-risk MDS patients, particularly elderly patients who are not candidates for intensive regimens. Pre-clinical data suggests that combining HMAs with immune checkpoint inhibitors in leukemia and other malignancies may enable synergistic activity and facilitate immunotherapy (Figure 2)[93–98]. HMAs and histone deacetylase (HDAC) inhibitors upregulate a variety of genes, including previously silenced leukemia antigens that could serve as targets for AML-specific immune responses. For example, treatment of AML cells with azacitidine, the HDAC inhibitor valproic acid, or the novel HMA SGI-110 induces expression of immunogenic cancer/testis antigens (CTAs) such as NY-ESO-1 and MAGE-A, and MAGE-specific cytotoxic T-lymphocyte (CTL) activity has been associated with major clinical responses[99–102]. Inhibitory immune checkpoint proteins including PD-L1, PD-L2, PD-1 and CTLA-4 can also be upregulated by treatment with HMAs[103]. Treatment with HMAs may therefore augment both tumor-specific immune responses by upregulating neo-antigens, as well as impair tumor-specific immune responses by upregulation of immune checkpoint proteins. However, such epigenetic drug-mediated immune checkpoint upregulation also provides rationale to overcome tumor immune resistance by the use of a PD-1 pathway inhibitor[103].
Figure 2. Combination epigenetic and immunotherapy in AML.

Proposed mechanisms of response to AML blasts in the setting of azacitidine (AZA). Increased expression of endogenous retroviral elements (ERV), IFN signaling, neoantigen presentation on MHC, and PD-L1 expression are shown. Antibodies against PD-L1 (e.g. atezolizumab, avelumab) with epigenetic therapy as a potential combination to target AML blasts and drive clonal expansion of tumor-specific T cells. (Adapted with permission from Stahl M, Curr Oncol Rep (2019) 21: 37)
Animal models have previously demonstrated a rationale for combination HMA and immune checkpoint inhibition. In a mouse model of metastatic breast cancer resistance to combined checkpoint blockade, co-treatment with azacitidine and the HDAC inhibitor entinostat with immune checkpoint inhibitors markedly improved treatment responses, curing more than 80% of tumor-bearing mice[96]. Additional studies demonstrated that this combination reduced numbers of myeloid-derived suppressor cells (MDSCs), suggesting that immune modulating effects of HMAs may potentially facilitate immunotherapy. Similar findings were observed in a mouse model of ovarian cancer, where the combination of decitabine and anti-CTLA-4 promoted differentiation of naive T cells into effector T cells, prolonging CTL responses as well as overall survival[97]. Clinical data in relapsed AML after transplant has demonstrated that the combination of azacitidine and donor lymphocyte infusion could induce long-term remissions, further supporting the concept of HMA-mediated facilitation of tumor-specific immunity[104].
Daver and colleagues systematically evaluated this question in an open label phase II study treating 70 relapsed/refractory AML patients with azacitidine and nivolumab[107]. Here, they demonstrated an overall response rate of 33% which is favorable to historical controls with other HMA combinations ranging between 10-15% in this setting. Of note, HMA-naïve patients, in addition to high levels of marrow infiltrating CD3 T cells at baseline had more favorable outcomes. Biomarker analysis also identified upregulation of CTLA-4+ CD4+ T cells (presumably Tregs) in non-responding patients. Although these combinations are promising in concept, the question remains to which patients would this provide the most benefit? We described the overt immunosuppressive state that can be established by increasing levels of disease (e.g. pDC depletion, T cell exhaustion, abortive tolerance), and would propose that patients with MRD would be the optimal population for treatment with immunotherapy.
The Emerging Importance of MRD in AML
The presence of minimal residual disease (MRD) after chemotherapy has emerged as a powerful prognostic factor in AML[12,108–112]. To this day, there are no approved therapies for AML patients with MRD. Due to differences in disease biology as well as patient age, the prognosis for older adults with MRD is particularly poor. Only 13% of AML patients over 60 with MRD after induction chemotherapy are alive and disease-free at five years[113]. In the setting of MRD, additional chemotherapy is often ineffective. It remains unclear if any post-remission therapy in older adults is beneficial[114], and even allogeneic stem cell transplantation (alloSCT) cannot rescue most adults with MRD after chemotherapy[12,108–111]. MRD positivity after consolidation chemotherapy in AML predicts early relapse and poorer outcomes with alloSCT, with one recent study indicating that transplant with MRD results in equivalently poor outcomes as transplant with active AML[12,108–111,113]. Data for monitoring AML MRD after transplant are limited, and there is currently no standard practice for post-transplant MRD monitoring. However, existing data indicate that the long-term prognosis for patients with AML MRD after alloSCT is also poor. AML MRD detection at day 100 post-transplant predicts relapse and poorer overall survival, and MRD detection at 1 year post-transplant may be an even more powerful negative prognostic marker[115–117]. In one recent study, patients who were MRD positive a year after alloSCT relapsed and died, while MRD-negative patients remained in CR[115].
Azacitidine with or without Checkpoint Blockade for Treatment of Minimal Residual Disease (MRD)
Single-agent Azacitidine has been used for the treatment of MRD in NPM1-mutant AML with molecular minimal residual disease[118]. Data are limited, but one case series describes 10 patients with normal karyotype AML and NPM1 mutations in first or second complete remission but with PCR detectable NPM1 consistent with MRD after intensive pretreatment with chemotherapy, allogeneic, or autologous HSCT. In this series, 7 out of 10 NPM1 MRD positive patients who underwent preemptive treatment with Azacitidine had a molecular response with an at least 1-log decrease in NPM1 MRD level. Five patients responded within 3 cycles of Azacitidine, and another 2 patients responded after cycle 4 and 5. However, some responses were only temporary. Three patients did achieve a sustained molecular complete response, but 2 out of these 3 patients underwent subsequent alloSCT. Intriguingly, 3 out of the 7 responders had previously undergone prior allogeneic SCT. Of the 3 patients with NPM1 MRD after prior allogeneic SCT, 1 achieved complete molecular response with clearance of MRD after 4 cycles, suggesting a potential enhancement of graft versus leukemia effect. Importantly, Azacitidine was well tolerated in this population, with reversible neutropenia and thrombocytopenia noted as the most frequent serious adverse events in 80% and 40% of patients, respectively.
Based on this and other data, Azacitidine has been used for treatment of MRD following alloSCT[119,120]. In the phase II RELapse prevention with AZAcitidine trials (RELAZA and RELAZA2), patients who experienced a decrease in CD34+ donor chimerism in the peripheral blood to values below 80% without concurrent hematologic relapse were treated with four cycles of Azacitidine. These studies demonstrated that pre-emptive therapy with azacitidine can prevent or substantially delay relapse in MRD+ patients. Azacitidine in this setting is well tolerated, with reversible grade ¾ neutropenia and thrombocytopenia and no significant exacerbations of GVHD. Lower-dose azacitidine has also been studied as maintenance therapy in AML patients during the first year after allogeneic transplant, with one study in 37 patients indicating that induction of a CD8+ T-cell response was associated with a reduced risk of relapse and improved relapse-free survival[121]. HMA treatment has also been previously shown to expand Tregs helping reduce GVHD while preserving this potential CD8+ T cell mediated GVL effect[122].
Several other post-transplant approaches with immune modulating agents have also been explored. Firstly, the use of the tyrosine kinase inhibitor, sorafenib, has been shown in pre-clinical models to increase levels of IL-15 and improve the phenotype (high levels of BCL-2 and decreased PD-1 levels) and function of leukemia-specific CD8 T cells. This increase in IL-15 levels is also mirrored in patients with FLT3-ITD+ AML responding to sorafenib post-alloHCT[123]. Additional studies with next-generation FLT3 inhibitors are ongoing. Secondly, for those patients without a targetable lesion such as FLT3 or IDH½, the use of the histone deacetylase (HDAC) inhibitor panobinostat has demonstrated impressive results in early phase studies [124]. While preclinical data support immunomodulatory activity and enhanced leukemia-specific activity with HDAC inhibitors, corollary studies are lacking from these early phase trials. Finally, the combination of HMA with the immune modulatory drug (IMiD) lenalidomide led to clinical responses in approximately half of patients treated at the recommended dose (25 mg daily with AZA 75 mg/m2 for 7 days)[125]. This did not, however, reverse T cell dysfunction in this population suggesting lenalidomide exerts its effects through other mechanisms.
In further support of a prominent role of immune-mediated control of AML both pre- and post-transplant, it was recently demonstrated that there is a significant downregulation of several MHC class II genes in patients who relapse following alloHCT[126]. This distinct mechanism of resistance has striking parallels to the recently described loss of MHC heterozygosity or beta-2-microglobulin (B2M) and immune escape in solid tumors during evolution or while receiving checkpoint blockade[127,128], supporting use of these novel agents in the treatment of AML.
Although checkpoint blockade could be a promising treatment approach for AML, we suggest that the patient population and specific immunotherapeutic agents will need to be optimized. First, lack of significant efficacy in alloHCT as disease burden increases suggest that PD-1/PD-L1 treatment in this same cohort may be ineffective secondary to the lack of DCs available for efficient cross-presentation in the setting of measurable disease. We propose that in order to overcome the abortive tolerance early on in the disease process secondary to engagement of CD8 T cells by expanding blasts, a more optimal approach may be to combine checkpoint blockade with HMA in the setting of MRD (Figure 2). This also provides a therapeutic option for many elderly patients unfit for further chemotherapy or allograft. This may also be an alternative treatment strategy for patients post-alloSCT who demonstrate evidence of disease reemergence, loss of donor chimerism, and need for additional therapies in combination with HMA or DLI.
Conclusion
We have much to learn regarding the use of immune modulating drugs for the treatment of patients with AML. Building on the expanding knowledge of immune suppression and dysfunction in AML patients remains a priority in the field. While we continue to rely on alloSCT as a cornerstone of treatment for patients with high-risk AML, it is reasonable to strive for alternatives that also engage a polyclonal immune response against chemotherapy-resistant cells. Cellular therapy will likely continue to play an important role, while the use of off-the-shelf NK cells or CAR-T cells becomes a more translatable approach. Although vaccine therapy remains an attractive concept in engaging both T- and B-cell mediated immunity following induction therapy, larger and more definitive trials are needed. Passive humoral immunity through the use of monoclonal antibodies will likely remain an active area of investigation in the field, given the ease at which these reagents can be generated and long track record of safety in hematologic malignancies. As novel cell-surface targets continue to be identified, these will need to be tested in early phase trials and hopefully moved to the upfront setting with combination chemotherapy. In the near future, larger trials testing immune checkpoint blockade alone or in combination with HMAs will be completed and we await these results. Evaluation of correlative studies from these patients remains critical as we attempt to better and understand and improve AML immunotherapy. Finally, the adverse prognostic significance of MRD remains an unmet need for therapy. We propose that AML MRD serves as an ideal target for combination therapies including HMA and checkpoint inhibition.
Acknowledgments
This work is supported by the American Society of Hematology Research Training Award for fellows and in part by grant # UL1 TR001866 from the National Center for Advancing Translational Sciences (NCATS, National Institutes of Health (NIH) Clinical and Translational Science Award (CTSA) program.
Footnotes
The authors report no conflict of interests.
References
- 1.Schiller GJ. High-risk acute myelogenous leukemia: treatment today ... and tomorrow. Hematology Am Soc Hematol Educ Program 2013;2013:201–208. [DOI] [PubMed] [Google Scholar]
- 2.Patel JP, Gonen M, Figueroa ME, et al. . Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N Engl J Med 2012;366:1079–1089. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Weinberg OK, Ohgami RS, Ma L, et al. . Acute myeloid leukemia with monosomal karyotype: morphologic, immunophenotypic, and molecular findings. Am J Clin Pathol 2014;142:190–195. [DOI] [PubMed] [Google Scholar]
- 4.Cytogenetic Mrozek K., molecular genetic, and clinical characteristics of acute myeloid leukemia with a complex karyotype. Semin Oncol 2008;35:365–377. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Bochtler T, Frohling S, Kramer A. Role of chromosomal aberrations in clonal diversity and progression of acute myeloid leukemia. Leukemia 2015. [DOI] [PubMed] [Google Scholar]
- 6.Burnett A, Wetzler M, Lowenberg B. Therapeutic advances in acute myeloid leukemia. J Clin Oncol 2011;29:487–494. [DOI] [PubMed] [Google Scholar]
- 7.Ossenkoppele G, Lowenberg B. How I treat the older patient with acute myeloid leukemia. Blood 2015;125:767–774. [DOI] [PubMed] [Google Scholar]
- 8.Klimek VM. Recent advances in the management of therapy-related myelodysplastic syndromes and acute myeloid leukemia. Curr Opin Hematol 2013;20:137–143. [DOI] [PubMed] [Google Scholar]
- 9.O’Donnell MR, Abboud CN, Altman J, et al. . Acute myeloid leukemia. J Natl Compr Canc Netw 2012;10:984–1021. [DOI] [PubMed] [Google Scholar]
- 10.Parmar S, Ritchie DS. Allogeneic transplantation as anticancer immunotherapy. Curr Opin Immunol 2014;27:38–45. [DOI] [PubMed] [Google Scholar]
- 11.Duval M, Klein JP, He W, et al. . Hematopoietic stem-cell transplantation for acute leukemia in relapse or primary induction failure. J Clin Oncol 2010;28:3730–3738. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Araki D, Wood BL, Othus M, et al. . Allogeneic Hematopoietic Cell Transplantation for Acute Myeloid Leukemia: Time to Move Toward a Minimal Residual Disease-Based Definition of Complete Remission? J Clin Oncol 2016;34:329–336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Weiden PL, Flournoy N, Thomas ED, et al. . Antileukemic Effect of Graft-versus-Host Disease in Human Recipients of Allogeneic-Marrow Grafts. New England Journal of Medicine 1979;300:1068–1073. [DOI] [PubMed] [Google Scholar]
- 14.Porter DL, Roth MS, McGarigle C, Ferrara JL, Antin JH. Induction of graft-versus-host disease as immunotherapy for relapsed chronic myeloid leukemia. The New England journal of medicine 1994;330:100–106. [DOI] [PubMed] [Google Scholar]
- 15.Norde WJ, Overes IM, Maas F, et al. . Myeloid leukemic progenitor cells can be specifically targeted by minor histocompatibility antigen LRH-1-reactive cytotoxic T cells. Blood 2009;113:2312–2323. [DOI] [PubMed] [Google Scholar]
- 16.Petersdorf EW. Optimal HLA matching in hematopoietic cell transplantation. Current opinion in immunology 2008;20:588–593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Montagna D, Maccario R, Locatelli F, et al. . Emergence of antitumor cytolytic T cells is associated with maintenance of hematologic remission in children with acute myeloid leukemia. Blood 2006;108:3843–3850. [DOI] [PubMed] [Google Scholar]
- 18.Reichardt VL, Brossart P. Current status of vaccination therapy for leukemias. Current hematology reports 2005;4:73–76. [PubMed] [Google Scholar]
- 19.Molldrem JJ. Vaccination for leukemia. Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation 2006;12:13–18. [DOI] [PubMed] [Google Scholar]
- 20.Marijt WAE, Heemskerk MHM, Kloosterboer FM, et al. . Hematopoiesis-restricted minor histocompatibility antigens HA-1- or HA-2-specific T cells can induce complete remissions of relapsed leukemia. Proceedings of the National Academy of Sciences of the United States of America 2003;100:2742–2747. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Ma Q, Wang C, Jones D, et al. . Adoptive transfer of PR1 cytotoxic T lymphocytes associated with reduced leukemia burden in a mouse acute myeloid leukemia xenograft model. Cytotherapy 2010;12:1056–1062. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Keilholz U, Letsch A, Busse A, et al. . A clinical and immunologic phase 2 trial of Wilms tumor gene product 1 (WT1) peptide vaccination in patients with AML and MDS. Blood 2009;113:6541–6548. [DOI] [PubMed] [Google Scholar]
- 23.Ho VT, Vanneman M, Kim H, et al. . Biologic activity of irradiated, autologous, GM-CSF-secreting leukemia cell vaccines early after allogeneic stem cell transplantation. Proceedings of the National Academy of Sciences of the United States of America 2009;106:15825–15830. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Uttenthal B, Martinez-Davila I, Ivey A, et al. . Wilms’ Tumour 1 (WT1) peptide vaccination in patients with acute myeloid leukaemia induces short-lived WT1-specific immune responses. British Journal of Haematology 2014;164:366–375. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Buckley SA, Walter RB. Antigen-specific immunotherapies for acute myeloid leukemia. Hematology / the Education Program of the American Society of Hematology. American Society of Hematology. Education Program 2015;2015:584–595. [DOI] [PubMed] [Google Scholar]
- 26.Zhang L, Chen X, Liu X, et al. . CD40 ligation reverses T cell tolerance in acute myeloid leukemia. Journal of Clinical Investigation 2013;123:1999–2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Kline DE, Fosco D, Chen X, Kline J. CD8α+ dendritic cells dictate immune responses against murine AML. Journal for ImmunoTherapy of Cancer 2013;1:P158. [Google Scholar]
- 28.Rock Kenneth L, Shen L. Cross-presentation: underlying mechanisms and role in immune surveillance. Immunological Reviews 2005;207:166–183. [DOI] [PubMed] [Google Scholar]
- 29.Mohty M, Jarrossay D, Lafage-Pochitaloff M, et al. . Circulating blood dendritic cells from myeloid leukemia patients display quantitative and cytogenetic abnormalities as well as functional impairment. Blood 2001;98:3750–3756. [DOI] [PubMed] [Google Scholar]
- 30.Xiao W, Goldberg AD, Famulare C, et al. . Loss of plasmacytoid dendritic cell differentiation is highly predictive for post-induction measurable residual disease and inferior outcomes in acute myeloid leukemia. Haematologica 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Williams P, Basu S, Garcia-Manero G, et al. . The distribution of T-cell subsets and the expression of immune checkpoint receptors and ligands in patients with newly diagnosed and relapsed acute myeloid leukemia. Cancer 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Curran E, Chen X, Corrales L, et al. . STING Pathway Activation Stimulates Potent Immunity against Acute Myeloid Leukemia. Cell Rep 2016;15:2357–2366. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Knaus HA, Berglund S, Hackl H, et al. . Signatures of CD8+ T cell dysfunction in AML patients and their reversibility with response to chemotherapy. JCI Insight 2018;3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Schnorfeil FM, Lichtenegger FS, Emmerig K, et al. . T cells are functionally not impaired in AML: increased PD-1 expression is only seen at time of relapse and correlates with a shift towards the memory T cell compartment. J Hematol Oncol 2015;8:93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Kronig H, Kremmler L, Haller B, et al. . Interferon-induced programmed death-ligand 1 (PD-L1/B7-H1) expression increases on human acute myeloid leukemia blast cells during treatment. Eur J Haematol 2014;92:195–203. [DOI] [PubMed] [Google Scholar]
- 36.Chen X, Liu S, Wang L, Zhang W, Ji Y, Ma X. Clinical significance of B7-H1 (PD-L1) expression in human acute leukemia. Cancer Biol Ther 2008;7:622–627. [DOI] [PubMed] [Google Scholar]
- 37.Salih HR, Wintterle S, Krusch M, et al. . The role of leukemia-derived B7-H1 (PD-L1) in tumor-T-cell interactions in humans. Exp Hematol 2006;34:888–894. [DOI] [PubMed] [Google Scholar]
- 38.Yang H, Bueso-Ramos C, DiNardo C, et al. . Expression of PD-L1, PD-L2, PD-1 and CTLA4 in myelodysplastic syndromes is enhanced by treatment with hypomethylating agents. Leukemia 2014;28:1280–1288. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Krupka C, Kufer P, Kischel R, et al. . Blockade of the PD-1/PD-L1 axis augments lysis of AML cells by the CD33/CD3 BiTE antibody construct AMG 330: reversing a T-cell-induced immune escape mechanism. Leukemia 2016;30:484–491. [DOI] [PubMed] [Google Scholar]
- 40.Berthon C, Driss V, Liu J, et al. . In acute myeloid leukemia, B7-H1 (PD-L1) protection of blasts from cytotoxic T cells is induced by TLR ligands and interferon-gamma and can be reversed using MEK inhibitors. Cancer Immunol Immunother 2010;59:1839–1849. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Cortez MA, Ivan C, Valdecanas D, et al. . PDL1 Regulation by p53 via miR-34. J Natl Cancer Inst 2016;108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Zhang L, Gajewski TF, Kline J. PD-1/PD-L1 interactions inhibit antitumor immune responses in a murine acute myeloid leukemia model. Blood 2009;114:1545–1552. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Ahmad SM, Svane IM, Andersen MH. The stimulation of PD-L1-specific cytotoxic T lymphocytes can both directly and indirectly enhance antileukemic immunity. Blood Cancer J 2014;4:e230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Zhou Q, Munger ME, Veenstra RG, et al. . Coexpression of Tim-3 and PD-1 identifies a CD8+ T-cell exhaustion phenotype in mice with disseminated acute myelogenous leukemia. Blood 2011;117:4501–4510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Zhou Q, Munger ME, Highfill SL, et al. . Program death-1 signaling and regulatory T cells collaborate to resist the function of adoptively transferred cytotoxic T lymphocytes in advanced acute myeloid leukemia. Blood 2010;116:2484–2493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Armand P Immune checkpoint blockade in hematologic malignancies. Blood 2015. [DOI] [PubMed] [Google Scholar]
- 47.Curran E, Corrales L, Kline J. Targeting the innate immune system as immunotherapy for acute myeloid leukemia. Front Oncol 2015;5:83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Restifo NP, Dudley ME, Rosenberg SA. Adoptive immunotherapy for cancer : harnessing the T cell response. Nature Publishing Group 2012;12:269–281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Greiner J, Ono Y, Hofmann S, et al. . Mutated regions of nucleophosmin 1 elicit both CD4(+) and CD8(+) T-cell responses in patients with acute myeloid leukemia. Blood 2012;120:1282–1289. [DOI] [PubMed] [Google Scholar]
- 50.Rezvani K, Yong ASM, Mielke S, et al. . Lymphodepletion is permissive to the development of spontaneous T-cell responses to the self-antigen PR1 early after allogeneic stem cell transplantation and in patients with acute myeloid leukemia undergoing WT1 peptide vaccination following chemotherapy. Cancer immunology, immunotherapy : CII 2012;61:1125–1136. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Perna F, Berman SH, Soni RK, et al. . Integrating Proteomics and Transcriptomics for Systematic Combinatorial Chimeric Antigen Receptor Therapy of AML. Cancer Cell 2017;32:506–519. e505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Hofmann S, Schubert ML, Wang L, et al. . Chimeric Antigen Receptor (CAR) T Cell Therapy in Acute Myeloid Leukemia (AML). J Clin Med 2019;8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Knorr DA, Bachanova V, Verneris MR, Miller JS. Clinical utility of natural killer cells in cancer therapy and transplantation. Seminars in immunology 2014;26:161–172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Lichtenegger FS, Lorenz R, Gellhaus K, Hiddemann W, Beck B, Subklewe M. Impaired NK cells and increased T regulatory cell numbers during cytotoxic maintenance therapy in AML. Leukemia research 2014;38:964–969. [DOI] [PubMed] [Google Scholar]
- 55.Kiessling R, Klein E, Wigzell H. “Natural” killer cells in the mouse. I. Cytotoxic cells with specificity for mouse Moloney leukemia cells. Specificity and distribution according to genotype. Eur J Immunol 1975;5:112–117. [DOI] [PubMed] [Google Scholar]
- 56.Murphy WJ, Parham P, Miller JS. NK cells--from bench to clinic. Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation 2012;18:S2–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Ruggeri L, Capanni M, Urbani E, et al. . Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science 2002;295:2097–2100. [DOI] [PubMed] [Google Scholar]
- 58.Miller JS, Soignier Y, Panoskaltsis-Mortari A, et al. . Successful adoptive transfer and in vivo expansion of human haploidentical NK cells in patients with cancer. Blood 2005;105:3051–3057. [DOI] [PubMed] [Google Scholar]
- 59.Knorr DA, Bachanova V, Verneris MR, Miller JS. Clinical utility of natural killer cells in cancer therapy and transplantation. Seminars in Immunology 2014;26:161–172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Knorr DA, Kaufman DS. Pluripotent stem cell-derived natural killer cells for cancer therapy. Translational Research 2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Cooley S, Trachtenberg E, Bergemann TL, et al. . Donors with group B KIR haplotypes improve relapse-free survival after unrelated hematopoietic cell transplantation for acute myelogenous leukemia. Blood 2009;113:726–732. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Venstrom JM, Pittari G, Gooley TA, et al. . HLA-C-dependent prevention of leukemia relapse by donor activating KIR2DS1. N Engl J Med 2012;367:805–816. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Vey N, Bourhis J-H, Boissel N, et al. . A phase 1 trial of the anti-inhibitory KIR mAb IPH2101 for AML in complete remission. Blood 2012;120:4317–4323. [DOI] [PubMed] [Google Scholar]
- 64.Ustun C, Miller JS, Munn DH, Weisdorf DJ, Blazar BR. Regulatory T cells in acute myelogenous leukemia: is it time for immunomodulation? Blood 2011:5084–5095. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Mantovani A The growing diversity and spectrum of action of myeloid-derived suppressor cells. European journal of immunology 2010;40:3317–3320. [DOI] [PubMed] [Google Scholar]
- 66.Kusmartsev S, Nefedova Y, Yoder D, Gabrilovich DI. Antigen-specific inhibition of CD8+ T cell response by immature myeloid cells in cancer is mediated by reactive oxygen species. Journal of immunology (Baltimore, Md. : 1950) 2004;172:989–999. [DOI] [PubMed] [Google Scholar]
- 67.Ding Z-C, Lu X, Yu M, et al. . Immunosuppressive myeloid cells induced by chemotherapy attenuate antitumor CD4+ T-cell responses through the PD-1-PD-L1 axis. Cancer research 2014;74:3441–3453. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Zhou Q, Munger ME, Veenstra RG, et al. . Coexpression of Tim-3 and PD-1 identifies a CD8+ T-cell exhaustion phenotype in mice with disseminated acute myelogenous leukemia. Blood 2011;117:4501–4510. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Zhou Q, Munger ME, Highfill SL, et al. . Program death-1 signaling and regulatory T cells collaborate to resist the function of adoptively transferred cytotoxic T lymphocytes in advanced acute myeloid leukemia. Blood 2010;116:2484–2493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Davids MS, Kim HT, Bachireddy P, et al. . Ipilimumab for Patients with Relapse after Allogeneic Transplantation. N Engl J Med 2016;375:143–153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Simpson TR, Li F, Montalvo-Ortiz W, et al. . Fc-dependent depletion of tumor-infiltrating regulatory T cells co-defines the efficacy of anti-CTLA-4 therapy against melanoma. The Journal of Experimental Medicine 2013;210:1695–1710. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Blazar BR, Taylor PA, Panoskaltsis-Mortari A, Sharpe AH, Vallera DA. Opposing roles of CD28:B7 and CTLA-4:B7 pathways in regulating in vivo alloresponses in murine recipients of MHC disparate T cells. Journal of immunology (Baltimore, Md. : 1950) 1999;162:6368–6377. [PubMed] [Google Scholar]
- 73.Zhou J, Bashey A, Zhong R, et al. . CTLA-4 blockade following relapse of malignancy after allogeneic stem cell transplantation is associated with T cell activation but not with increased levels of T regulatory cells. Biology of blood and marrow transplantation : journal of the American Society for Blood and Marrow Transplantation 2011;17:682–692. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Rosenblatt J, Stone RM, Uhl L, et al. . Individualized vaccination of AML patients in remission is associated with induction of antileukemia immunity and prolonged remissions. Sci Transl Med 2016;8:368ra171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Ott PA, Hu Z, Keskin DB, et al. . An immunogenic personal neoantigen vaccine for patients with melanoma. Nature 2017;547:217–221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Mitterbauer M, Fritzer-Szekeres M, Mitterbauer G, et al. . Spontaneous remission of acute myeloid leukemia after infection and blood transfusion associated with hypergammaglobulinaemia. Annals of Hematology 1996;73:189–193. [DOI] [PubMed] [Google Scholar]
- 77.Greiner J, Ringhoffer M, Taniguchi M, et al. . Characterization of several leukemia-associated antigens inducing humoral immune responses in acute and chronic myeloid leukemia. International Journal of Cancer 2003;106:224–231. [DOI] [PubMed] [Google Scholar]
- 78.Gillissen MA, Kedde M, Jong Gd, et al. . AML-specific cytotoxic antibodies in patients with durable graft-versus-leukemia responses. Blood 2018;131:131. [DOI] [PubMed] [Google Scholar]
- 79.Gillissen MA, de Jong G, Kedde M, et al. . Patient-derived antibody recognizes a unique CD43 epitope expressed on all AML and has antileukemia activity in mice. Blood Advances 2017;1:1551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Lichtenegger FS, Krupka C, Haubner S, Kohnke T, Subklewe M. Recent developments in immunotherapy of acute myeloid leukemia. J Hematol Oncol 2017;10:142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Lesokhin AM, Callahan MK, Postow MA, Wolchok JD. On being less tolerant: enhanced cancer immunosurveillance enabled by targeting checkpoints and agonists of T cell activation. Sci Transl Med 2015;7:280sr281. [DOI] [PubMed] [Google Scholar]
- 82.Postow MA, Chesney J, Pavlick AC, et al. . Nivolumab and ipilimumab versus ipilimumab in untreated melanoma. N Engl J Med 2015;372:2006–2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Barbee MS, Ogunniyi A, Horvat TZ, Dang TO. Current Status and Future Directions of the Immune Checkpoint Inhibitors Ipilimumab, Pembrolizumab, and Nivolumab in Oncology. Ann Pharmacother 2015. [DOI] [PubMed] [Google Scholar]
- 84.Lesokhin AM, Callahan MK, Postow MA, Wolchok JD. On being less tolerant: enhanced cancer immunosurveillance enabled by targeting checkpoints and agonists of T cell activation. Science Translational Medicine 2015;7:280sr281. [DOI] [PubMed] [Google Scholar]
- 85.Tumeh PC, Harview CL, Yearley JH, et al. . PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 2014;515:568–571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Ansell SM, Lesokhin AM, Borrello I, et al. . PD-1 blockade with nivolumab in relapsed or refractory Hodgkin’s lymphoma. N Engl J Med 2015;372:311–319. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Boddu P, Kantarjian H, Garcia-Manero G, Allison J, Sharma P, Daver N. The emerging role of immune checkpoint based approaches in AML and MDS. Leuk Lymphoma 2018;59:790–802. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Lawrence MS, Stojanov P, Polak P, et al. . Mutational heterogeneity in cancer and the search for new cancer-associated genes. Nature 2013;499:214–218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Cancer Genome Atlas Research N. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med 2013;368:2059–2074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Rizvi NA, Hellmann MD, Snyder A, et al. . Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science 2015;348:124–128. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Snyder A, Makarov V, Merghoub T, et al. . Genetic basis for clinical response to CTLA-4 blockade in melanoma. N Engl J Med 2014;371:2189–2199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Le DT, Uram JN, Wang H, et al. . PD-1 Blockade in Tumors with Mismatch-Repair Deficiency. N Engl J Med 2015;372:2509–2520. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Heninger E, Krueger TE, Lang JM. Augmenting antitumor immune responses with epigenetic modifying agents. Front Immunol 2015;6:29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Chiappinelli KB, Strissel PL, Desrichard A, et al. . Inhibiting DNA Methylation Causes an Interferon Response in Cancer via dsRNA Including Endogenous Retroviruses. Cell 2015;162:974–986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Roulois D, Loo Yau H, Singhania R, et al. . DNA-Demethylating Agents Target Colorectal Cancer Cells by Inducing Viral Mimicry by Endogenous Transcripts. Cell 2015;162:961–973. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Kim K, Skora AD, Li Z, et al. . Eradication of metastatic mouse cancers resistant to immune checkpoint blockade by suppression of myeloid-derived cells. Proc Natl Acad Sci U S A 2014;111:11774–11779. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Wang L, Amoozgar Z, Huang J, et al. . Decitabine Enhances Lymphocyte Migration and Function and Synergizes with CTLA-4 Blockade in a Murine Ovarian Cancer Model. Cancer Immunol Res 2015;3:1030–1041. [DOI] [PubMed] [Google Scholar]
- 98.Daver N, Boddu P, Garcia-Manero G, et al. . Hypomethylating agents in combination with immune checkpoint inhibitors in acute myeloid leukemia and myelodysplastic syndromes. Leukemia 2018;32:1094–1105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Almstedt M, Blagitko-Dorfs N, Duque-Afonso J, et al. . The DNA demethylating agent 5-aza-2’-deoxycytidine induces expression of NY-ESO-1 and other cancer/testis antigens in myeloid leukemia cells. Leuk Res 2010;34:899–905. [DOI] [PubMed] [Google Scholar]
- 100.Srivastava P, Paluch BE, Matsuzaki J, et al. . Immunomodulatory action of SGI-110, a hypomethylating agent, in acute myeloid leukemia cells and xenografts. Leuk Res 2014;38:1332–1341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Goodyear O, Agathanggelou A, Novitzky-Basso I, et al. . Induction of a CD8+ T-cell response to the MAGE cancer testis antigen by combined treatment with azacitidine and sodium valproate in patients with acute myeloid leukemia and myelodysplasia. Blood 2010;116:1908–1918. [DOI] [PubMed] [Google Scholar]
- 102.Srivastava P, Paluch BE, Matsuzaki J, et al. . Induction of cancer testis antigen expression in circulating acute myeloid leukemia blasts following hypomethylating agent monotherapy. Oncotarget 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Orskov AD, Treppendahl MB, Skovbo A, et al. . Hypomethylation and up-regulation of PD-1 in T cells by azacytidine in MDS/AML patients: A rationale for combined targeting of PD-1 and DNA methylation. Oncotarget 2015;6:9612–9626. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Steinmann J, Bertz H, Wasch R, et al. . 5-Azacytidine and DLI can induce long-term remissions in AML patients relapsed after allograft. Bone Marrow Transplant 2015;50:690–695. [DOI] [PubMed] [Google Scholar]
- 105.Dear AE. Epigenetic Modulators and the New Immunotherapies. N Engl J Med 2016;374:684–686. [DOI] [PubMed] [Google Scholar]
- 106.Peng D, Kryczek I, Nagarsheth N, et al. . Epigenetic silencing of TH1-type chemokines shapes tumour immunity and immunotherapy. Nature 2015;527:249–253. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Daver N, Garcia-Manero G, Basu S, et al. . Efficacy, Safety, and Biomarkers of Response to Azacitidine and Nivolumab in Relapsed/Refractory Acute Myeloid Leukemia: A Non-randomized, Open-label, Phase 2 Study. Cancer Discov 2018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Terwijn M, van Putten WL, Kelder A, et al. . High prognostic impact of flow cytometric minimal residual disease detection in acute myeloid leukemia: data from the HOVON/SAKK AML 42A study. J Clin Oncol 2013;31:3889–3897. [DOI] [PubMed] [Google Scholar]
- 109.Walter RB, Buckley SA, Pagel JM, et al. . Significance of minimal residual disease before myeloablative allogeneic hematopoietic cell transplantation for AML in first and second complete remission. Blood 2013;122:1813–1821. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Kayser S, Walter RB, Stock W, Schlenk RF. Minimal Residual Disease in Acute Myeloid Leukemia-Current Status and Future Perspectives. Curr Hematol Malig Rep 2015. [DOI] [PubMed] [Google Scholar]
- 111.Anthias C, Dignan FL, Morilla R, et al. . Pre-transplant MRD predicts outcome following reduced-intensity and myeloablative allogeneic hemopoietic SCT in AML. Bone Marrow Transplant 2014;49:679–683. [DOI] [PubMed] [Google Scholar]
- 112.Ivey A, Hills RK, Simpson MA, et al. . Assessment of Minimal Residual Disease in Standard-Risk AML. N Engl J Med 2016;374:422–433. [DOI] [PubMed] [Google Scholar]
- 113.Buccisano F, Maurillo L, Piciocchi A, et al. . Minimal residual disease negativity in elderly patients with acute myeloid leukemia may indicate different postremission strategies than in younger patients. Ann Hematol 2015;94:1319–1326. [DOI] [PubMed] [Google Scholar]
- 114.Goldstone AH, Burnett AK, Wheatley K, et al. . Attempts to improve treatment outcomes in acute myeloid leukemia (AML) in older patients: the results of the United Kingdom Medical Research Council AML11 trial. Blood 2001;98:1302–1311. [DOI] [PubMed] [Google Scholar]
- 115.Bastos-Oreiro M, Perez-Corral A, Martinez-Laperche C, et al. . Prognostic impact of minimal residual disease analysis by flow cytometry in patients with acute myeloid leukemia before and after allogeneic hemopoietic stem cell transplantation. Eur J Haematol 2014;93:239–246. [DOI] [PubMed] [Google Scholar]
- 116.Bernal T, Diez-Campelo M, Godoy V, et al. . Role of minimal residual disease and chimerism after reduced-intensity and myeloablative allo-transplantation in acute myeloid leukemia and high-risk myelodysplastic syndrome. Leuk Res 2014;38:551–556. [DOI] [PubMed] [Google Scholar]
- 117.Diez-Campelo M, Perez-Simon JA, Perez J, et al. . Minimal residual disease monitoring after allogeneic transplantation may help to individualize post-transplant therapeutic strategies in acute myeloid malignancies. Am J Hematol 2009;84:149–152. [DOI] [PubMed] [Google Scholar]
- 118.Sockel K, Wermke M, Radke J, et al. . Minimal residual disease-directed preemptive treatment with azacitidine in patients with NPM1-mutant acute myeloid leukemia and molecular relapse. Haematologica 2011;96:1568–1570. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Platzbecker U, Wermke M, Radke J, et al. . Azacitidine for treatment of imminent relapse in MDS or AML patients after allogeneic HSCT: results of the RELAZA trial. Leukemia 2012;26:381–389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Platzbecker U, Middeke JM, Sockel K, et al. . Measurable residual disease-guided treatment with azacitidine to prevent haematological relapse in patients with myelodysplastic syndrome and acute myeloid leukaemia (RELAZA2): an open-label, multicentre, phase 2 trial. Lancet Oncol 2018;19:1668–1679. [DOI] [PubMed] [Google Scholar]
- 121.Craddock C, Jilani N, Siddique S, et al. . Tolerability and Clinical Activity of Post-Transplantation Azacitidine in Patients Allografted for Acute Myeloid Leukemia Treated on the RICAZA Trial. Biol Blood Marrow Transplant 2016;22:385–390. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Goodyear OC, Dennis M, Jilani NY, et al. . Azacitidine augments expansion of regulatory T cells after allogeneic stem cell transplantation in patients with acute myeloid leukemia (AML). Blood 2012;119:3361–3369. [DOI] [PubMed] [Google Scholar]
- 123.Mathew NR, Baumgartner F, Braun L, et al. . Sorafenib promotes graft-versus-leukemia activity in mice and humans through IL-15 production in FLT3-ITD-mutant leukemia cells. Nat Med 2018;24:282–291. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Bug G, Burchert A, Wagner E-M, et al. . Phase I/II study of the deacetylase inhibitor panobinostat after allogeneic stem cell transplantation in patients with high-risk MDS or AML (PANOBEST trial). Leukemia 2017;31:2523. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Craddock C, Slade D, De Santo C, et al. . Combination Lenalidomide and Azacitidine: A Novel Salvage Therapy in Patients Who Relapse After Allogeneic Stem-Cell Transplantation for Acute Myeloid Leukemia. J Clin Oncol 2019;37:580–588. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Christopher MJ, Petti AA, Rettig MP, et al. . Immune Escape of Relapsed AML Cells after Allogeneic Transplantation. N Engl J Med 2018;379:2330–2341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.McGranahan N, Rosenthal R, Hiley CT, et al. . Allele-Specific HLA Loss and Immune Escape in Lung Cancer Evolution. Cell 2017;171:1259–1271. e1211. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Zaretsky JM, Garcia-Diaz A, Shin DS, et al. . Mutations Associated with Acquired Resistance to PD-1 Blockade in Melanoma. N Engl J Med 2016;375:819–829. [DOI] [PMC free article] [PubMed] [Google Scholar]
